Antimicrobial Activity of Selected Antimicrobial

9 downloads 0 Views 467KB Size Report
The main mechanism of their antimicrobial activity is based on a ... Poland) and OxymaPure (Iris Biotech GmbH, Marktredwitz,. Germany). Every step was ...
Probiotics and Antimicrobial Proteins https://doi.org/10.1007/s12602-018-9444-5

Antimicrobial Activity of Selected Antimicrobial Peptides Against Planktonic Culture and Biofilm of Acinetobacter baumannii Maciej Jaśkiewicz 1

&

Damian Neubauer 1 & Kamil Kazor 1 & Sylwia Bartoszewska 1 & Wojciech Kamysz 1

# The Author(s) 2018

Abstract Acinetobacter baumannii is one of the most challenging pathogens, on account of its predisposition to develop resistance leading to severe, difficult-to-treat infections. As these bacteria are more usually isolated from nosocomial infections, the new therapeutic options are demanded. Antimicrobial peptides (AMPs) are compounds likely to find application in the treatment of A. baumannii. These compounds exhibit a wide spectrum of antimicrobial activity and were found to be effective against biofilm. In this study, eight AMPs, namely aurein 1.2, CAMEL, citropin 1.1., LL-37, omiganan, r-omiganan, pexiganan, and temporin A, were tested for their antimicrobial activity. A reference strain of A. baumannii ATCC 19606 was used. Antimicrobial assays included determination of the minimum inhibitory concentration and the minimum biofilm eradication concentration. Considering the fact that the majority of A. baumannii infections are associated with mechanical ventilation and the use of indwelling devices, the activity against biofilm was assessed on both a polystyrene surface and tracheal tube fragments. In addition, cytotoxicity (HaCaT) was determined and in vitro selectivity index was calculated. Keywords Acinetobacter baumannii . Antibiotics . Antimicrobial peptides . Antimicrobial susceptibility testing . Biofilm . Peptide drugs . Tracheal tubes

Abbreviations AMPs Antimicrobial peptides ATCC American Type Culture Collection CFU Colony-forming units DCM Dichloromethane DIC N,N′-diisopropylcarbodiimide DMF N,N-dimethylformamide DMSO Dimethyl sulfoxide ESI-MS Electrospray ionization mass spectrometry Fmoc N-9-fluorenylmethoxycarbonyl HPLC High-performance liquid chromatography MBC Minimum bactericidal concentration MBEC Minimum biofilm eradication concentration MBECtt Minimum biofilm eradication concentration on tracheal tube fragments MHA Mueller-Hinton agar MHB Mueller-Hinton broth

* Maciej Jaśkiewicz [email protected] 1

Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland

MIC MTT PBS RP-HPLC SI TFA TIS

Minimum inhibitory concentration 3-(4,5-Dimethyl-2-thiazolyl)-2,5diphenyl-2-H-tetrazoluim bromide Phosphate-buffered saline Reversed-phase liquid chromatography Selectivity index Trifluoroacetic acid Triisopropylosilane

Introduction The Acinetobacter bacteria are currently one of the major causes of nosocomial infections while Acinetobacter baumannii is considered to be the major pathogen, owing to its predisposition to develop resistance [1]. These aerobic, Gram-negative bacteria have been routinely isolated form nosocomial infections, notably from patients of intensive care units [2]. These infections are frequently severe and difficult to treat, on account of their correlation with inter-regional spread and local occurrence of A. baumannii resistant to carbapenems [3]. This fact is still more alarming since these βlactam antibiotics are considered to be the last-line remedy

Probiotics & Antimicro. Prot.

used in the treatment of patients infected with multi-resistant Gram-negative bacteria [4]. The A. baumannii causes a wide variety of infections, but most of them are associated with the respiratory tract and the use of indwelling devices. Therefore, hospital-acquired pneumonia is the most frequent clinical condition connected with these bacteria, particularly associated with patients receiving the mechanical ventilator assistance [5, 6]. In addition, A. baumannii can also cause blood stream infections, wound and urinary tract infections, and meningitis [7, 8]. Treatment of those infections, if caused by multidrugresistant bacteria, involves the use of combinations of such agents as colistin, polymyxin B, and tigecycline. However, the safety of these approaches, even if successful, has not yet been well documented [9–11]. In this situation, the search for new, effective antimicrobials is demanded. Antimicrobial peptides are an interesting class of compounds that can be an alternative to conventional antibiotics. These endogenic molecules are widely distributed in nature, mainly as a part of innate immunity of organisms [12]. They target a broad spectrum of pathogens (bacteria, fungi, protozoa, and viruses) and are associated with triggering and coordinating multiple components of innate and immune adaptive systems [13–16]. Owing to these properties, antimicrobial peptides (AMPs) have attracted much attention of researchers from many scientific groups worldwide. Generally, AMPs are 11–50 amino acid residue long, amphipathic molecules with net positive charge [17, 18]. The main mechanism of their antimicrobial activity is based on a non-receptor-mediated microbial membrane disruption. However, they can also lead to microbial cell death through other mechanisms such as inhibition of protein/ DNA or cell wall synthesis or induction of apoptosis/necrosis. Owing to the membrane-associated activity, they could also exhibit a low potency to trigger antimicrobial resistance [19]. The aim of this study was to evaluate the activity of eight AMPs, namely aurein 1.2, CAMEL, citropin 1.1., LL-37, omiganan, r-omiganan, pexiganan, and temporin A, against reference a strain of A. baumannii ATCC 19606. The microbiological studies involved the determination of MIC, MBC, and MBEC. Interestingly, the antibiofilm activity was evaluated both on a polystyrene surface and tracheal tube fragments. The results were correlated with those of conventional antibiotics: ciprofloxacin, erythromycin, gentamicin, piperacillin, rifampicin, and tigecycline. Cytotoxicity of tested peptides was determined by MTT assay.

Table 1

Peptides used in this study

Name

Sequence

Aurein 1.2 CAMEL Citropin 1.1 LL-37

GLFDIIKKIAESF-NH2 KWKLFKKIGAVLKVL-NH2 GLFDVIKKVASVIGGL-NH2 LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES

Omiganan r-Omiganan Pexiganan Temporin A

ILRWPWWPWRRK-NH2 KRRWPWWPWRLI-NH2 GIGKFLKKAKKFGKAFVKILKK-NH2 FLPLIGRVLSGIL-NH2

the Fmoc group was carried out in a 20% (v/v) piperidine (Merck, Darmstadt, Germany) solution in DMF (Honeywell, Seelze, Germany) for 15 min. Acylation with protected amino acid was conducted in a DMF/DCM solution (Chempur, Piekary Slaskie, Poland) with coupling agents for 1.5 h using a threefold molar excess of DIC (Peptideweb, Zblewo, Poland) and OxymaPure (Iris Biotech GmbH, Marktredwitz, Germany). Every step was preceded by rinsing the resin and running the chloranil test. Peptides were cleaved from the resin using one of the mixtures: (A) TFA (Apollo Scientific, Denton, UK), phenol, TIS (Sigma-Aldrich, St. Louise, MO, USA), and water (92.5:2.5:2.5:2.5 v/v); (B) TFA, triisopropylsilane, and water (95:2.5:2.5 v/v). Mixture (A) was used with peptides containing a tryptophan residue, whereas mixture (B) with the remaining peptides. Crude peptides were precipitated with cold diethyl ether (Chempur, Piekary Slaskie, Poland) and lyophilized. Subsequently, the peptides were purified by RP-HPLC with LP-chrom software (Lipopharm.pl, Poland). Purifications were carried out on a Phenomenex Gemini-NX C18 column (21.20 × 100 mm, 5.0-μm particle size, 110-Å pore size). UV detection at 214 nm was used, and crude peptides were eluted with a linear 10–70% acetonitrile gradient in deionized water over 90 min at room temperature. The mobile phase flow rate was 10.0 mL/min. Both eluents contained 0.1% (v/v) of TFA. Fractions were analyzed on a Waters X-Bridge Shield RP-18 column (4.6 × 150 mm, 3.5-μm particle size, 130-Å pore size) with UV detection at 214 nm. Pure fractions (> 95%, by HPLC analysis) were collected and lyophilized. The identity of all compounds was confirmed by mass spectrometry (ESIMS).

Cultivation of Organisms

Materials and Methods Peptide Synthesis Peptides used in this study (Table 1) were synthesized manually by solid-phase method using Fmoc chemistry on polystyrene resin modified by a Rink amide linker. Deprotection of

The A. baumannii ATCC 19606 strain was acquired from the American Type Culture Collection and prepared in line with the manufacturer’s instructions. The culture was stored in Roti®-Store Cryo-Vials (Carl Roth GmbH, Karlsruhe, Germany) at − 20 °C. Before the tests, the bead with cryoprotected bacteria was transferred into fresh MHB (Biocorp,

Probiotics & Antimicro. Prot.

Warsaw, Poland) and incubated for 24 h at 37 °C. Then, the culture was seeded on MHA plates and incubated as just mentioned. The agar cultures prepared in this way were used for further experiments. Cell densities for all assays were adjusted spectrophotometrically (Multiskan™ GO Microplate Spectrophotometer, Thermo Scientific) at 600 nm.

Activity Against Planktonic Forms of Bacteria The MICs and MBCs were determined with the reference to the Clinical and Laboratory Standards Institute guidelines [20]. MICs were determined by broth microdilution method. For this purpose, initial inoculums of bacteria (0.5 × 105 CFU/ mL) in MHB were exposed to a series of concentrations (0.5– 256 μg/mL) of the compounds and incubated for 18 h at 37 °C. The experiments were conducted on 96-well microtiter polystyrene plates with the final volume of 100 μL. The MIC was taken as the lowest drug concentration at which a noticeable growth of microorganisms was inhibited. In parallel, the MIC wells, two above and one below (positive control), were cultured on a MHA plate. The lowest concentration of test compounds that did not show any visible growth of bacteria on the solid medium after 24 h of incubation at 37 °C were defined as MBC. All experiments were conducted in triplicate.

Activity Against Biofilm The antibiofilm activity was determined on 96-well flat-bottomed microtiter polystyrene plates and 0.5-cm fragments of tracheal tubes (RIM-75, ZARYS International Group, Zabrze, Poland), with resazurin (7-hydroxy-3H-phenoxazin-3-one 10-oxide) as a cell viability reagent. The first part included assay on 96-well plates. To do this, the plates were filled with 100 μL of the initial inoculums of bacteria (0.5 × 107 CFU/ mL) in the MHB and incubated for 24 h at 37 °C. Then, the wells were rinsed three times with PBS to remove nonadhered cells and the fresh medium with a series of concentrations (0.5–256 μg/mL) of the compounds was added. After 24 h of incubation, 20 μL of resazurin (4 mg/mL) was added to each well and the MBEC values were read. The assay on tracheal tube fragments was conducted using 24-well polystyrene plates. Briefly, the tracheal tube fragments were dipped in 1 mL of the bacterial inoculum (0.5 × 107 CFU/ mL) and incubated at 37 °C. After 24 h of incubation, the test fragments were rinsed three times with PBS and transferred into other wells containing the fresh medium with a series of concentrations of the test compounds. Subsequently, the plates were incubated again for 24 h at 37 °C. Then, 200 μL of resazurin was added and the MBEC was read. All experiments were conducted in triplicate.

MTT Assay To evaluate the cytotoxicity of the test peptides (IC50), the classic MTT assay on 96-well plates was performed for human keratinocytes (HaCaT) which were acquired from the ATCC. The assay utilizes colorimetric determination of the cell metabolic activity and the color intensity reflects the number of live cells that can be measured spectrophotometrically. The cell line was cultured in Dulbecco’s modified Eagle Medium (Invitrogen) supplemented with 10% fetal bovine serum (v/v), 100 units/mL of penicillin, 100 μg/mL of streptomycin, and 2 mM L-glutamine and was kept at 37 °C in a humidified 5% CO2 incubator. Briefly, a day after plating of 500 cells per well, a series of concentrations (0.5–500 μg/mL) of the test compounds were applied. DMSO was added to the control cells at a final concentration of 1.0% (v/v), which was related to the maximal concentration of the solvent compounds used in the experiment. After 24 h of incubation at 37 °C (humidified 5% CO2 incubator) with the peptides, a medium containing 1 mg/mL of MTT was added to the wells up to a final concentration of 0.5 mg/mL. Subsequently, the plates were incubated at 37 °C for 4 h. Then, the medium was aspirated, and the formazan product was solubilized with DMSO. The background absorbance at 630 nm was subtracted from that at 570 nm for each well (Epoch, BioTek Instruments, USA). Six replicates were conducted for each concentration. All experiments were repeated at least twice and the resulting IC50 values were calculated with a GraFit 7 software (v. 7.0, Erithacus, Berkley, CA, USA).

Results Antimicrobial Assay Eight different AMPs and six conventional antibiotics were tested against the reference strain of A. baumannii ATCC 19606. The results indicate that the peptides exhibit a high activity against planktonic forms of the bacteria (Table 2). The most effective was CAMEL and pexiganan with MIC and MBC values of 2 and 4 μg/mL, respectively. The next was LL-37 with an order of magnitude lower activity and citropin 1.1 with MIC and MBC values of 16 μg/mL. Interestingly, the identical activities exhibited aurein 1.2 and r-omiganan, the retro analog of omiganan, which turned out to be stronger than the parent compound (for which the MICs and MBCs were 32 and 64 μg/mL, respectively). Temporin A exhibited the highest minimum inhibitory and bactericidal concentrations of 128 μg/mL. In conclusion, the antimicrobial activity of the peptides decreased in the order: CAMEL, pexiganan > LL-37 > aurein 1.2, citropin 1.1, r-omiganan > omiganan > temporin A. Among the antibiotics tested, the most antimicrobially active was ciprofloxacin with MICs

Probiotics & Antimicro. Prot. Table 2 Antimicrobial activity of the test compounds [μg/mL] against A. baumannii ATCC 19606 Compound

MIC

MBC

MBEC

MBECtt

Ciprofloxacin

0.5

1

2

1

Erythromycin Gentamicin

32 16

32 32

32 32

16 32

Piperacillin

64

128

> 512

> 512

Rifampicin Tetracycline

4 2

4 4

8 16

4 4

Aurein 1.2

16

16

64

32

CAMEL Citropin 1.1

2 16

4 16

128 256

64 32

LL-37 Omiganan

4 32

8 64

512 256

128 128

r-Omiganan

16

16

256

128

Pexiganan Temporin A

2 128

4 128

256 512

32 256

and MBCs of 0.5 and 1 μg/mL, respectively. With rifampicin and tetracycline, these quantities ranged between 2 and 4 μg/ mL, while with erythromycin and gentamicin between 16 and 32 μg/mL. Piperacillin was the least effective antibiotic with MICs and MBCs of 64 and 128 μg/mL, respectively. In general, the antimicrobial activity of antibiotics decreased in the order: ciprofloxacin > tetracycline, rifampicin > gentamicin, erythromycin > piperacillin.

Antibiofilm Activity Antibiofilm studies and the results of MBEC indicate that eradication of biofilm from tracheal tubes occurred as a rule at lower concentrations than that of the polystyrene surface of microtiter plates (Table 2). However, the tested AMPs appear to exhibit a lower antibiofilm activity than did conventional antibiotics. The most active compound was ciprofloxacin with MBEC values for polystyrene and tracheal tubes of 2 and 1 μg/mL, respectively. Interestingly, erythromycin eradication concentrations were identical to those of MIC and MBC and by an order of magnitude lower for tracheal tubes. The antibiofilm activity of gentamicin and rifampicin appears to be almost equal for planktonic cultures while with tetracycline, this activity was found to be threefold lower. Among all the test compounds, only piperacillin failed to eradicate the biofilm at any concentration. The most active peptide was aurein 1.2 with up to twice as high MBEC than MIC. On the other hand, the largest difference between MICs and MBECs was found for CAMEL (the sixfold lower activity), pexiganan, and LL-37 (the sevenfold lower activity). What is more, identical MBECs were found for omiganan and its retro analog. Temporin A was up to twice less active against biofilm, but the concentrations were relatively high (512 and

256 μg/mL for biofilm on the polystyrene and tracheal tubes, respectively). In general, the MBECtt were either lower (up to threefold in magnitude) or equal to MBEC for all of the test compounds (antibiotics and peptides).

Cytotoxicity Evaluation of cytotoxicity of the majority of peptides used in this study was conducted by us previously and the present analyses were run for omiganan and r-omiganan only [21]. Both the previous and actual results were used for calculation of SI (Table 3). Based on the cumulative set of the IC50 values, it can be stated that the majority of peptides exhibited relatively high toxicity against HaCaT cell line. The IC50 values ranged between 1.05 and 79.39 μg/mL and for half of them were lower than the MIC (the exception was found for CAMEL, LL-37, omiganan, and r-omiganan). Interestingly, the IC50 for LL-37 was nearly three times lower than the MIC value (SI = 2.93).

Discussion Recent scientific reports highlight the problem of an increasing resistance to antibiotics and global spread of multidrugresistant bacteria. For this group, a special term of ESKAPE pathogens has been proposed, which strictly refers to specific species namely, Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter [22]. As these bacteria are frequently isolated from patients with severe, difficult-to-treat infections, new therapeutic options are needed. Moreover, the research on new antimicrobial compounds should be focused on this particular group of pathogens with orientation on reference as well as clinical isolates and the activity against biofilm. It is commonly known that survival potential of bacteria is enhanced by biofilm formation, but this feature is not always taken into account in the clinic. In fact, the susceptibility profile of bacteria is routinely determined for Table 3

IC50 values and SI of the peptides

Compound

IC50

MIC

SI (IC50//MIC)

CAMEL Citropin 1.1 LL-37 Omiganan r-Omiganan Pexiganan Temporin A

3.32* 3.02* 11.75* 79.39 29.51 1.05* 7.91*

2 16 4 32 16 2 128

1.66 0.18 2.93 2.48 1.84 0.52 0.06

*

These analyses were performed by our group in the previous study [21]

Probiotics & Antimicro. Prot.

planktonic cultures. However, biofilm can be described as a cellular conglomerate attached to a surface (both biotic and abiotic) enclosed in a matrix built up with extracellular polymeric substance (EPS) [23, 24]. As compared to the freely suspended planktonic cultures, this structure is 10 up to 1000 times more resistant to antimicrobials. Particularly noteworthy is the fact that the biofilm is responsible for over 80% of microbial infections in the body. Moreover, biofilmproducing bacteria have a capability to colonize indwelling devices, thus leading to infections [24–26]. A. baumannii is one of the strains that causing medical device colonization becomes one of the major sources of systemic dissemination [27]. AMPs as compounds with great antibiofilm potential can find application in the treatment of A. baumannii infections [28–32]. In this study, eight AMPs, namely, aurein 1.2, CAMEL, citropin 1.1., LL-37, pexiganan, omiganan, romiganan, and temporin A, were tested using reference strain of A. baumannii ATCC 19606. Furthermore, the antibiofilm activity was also determined for tracheal tube fragments. This approach is quite unique since medical devices are used more likely for the analysis of bacteria or their isolation [33–35]. As a matter of fact, the growth of A. baumannii on tracheal tubes is one of the major sources of colonization in lungs and bacterial pneumonia [36, 37]. Among the tested peptides, CAMEL and pexiganan displayed the highest activity with MIC values of 2 μg/mL. These results are consistent with other studies conducted on clinical strains of A. baumannii. For example, Giacometti et al. conducted a research on in vitro activity of cecropin A, melittin, and CAMEL [CA(1–7)M(2– 9)NH2], alone and in combination with antibiotics, against 32 nosocomial isolates of A. baumannii [38]. The MICs and MBCs ranged between 0.25–8 and 0.5–16 μg/mL, respectively. In spite of the fact that the peptide was found to be very active with the potential in adjuvant therapy with other antibiotics, the authors suggested that further studies on safety determination had to be conducted. Pexiganan is another peptide that was previously tested against clinical isolates. For instance, in the study conducted by Ge et al., susceptibility profiles of a number of strains were determined [39]. The distribution of the MICs ranged between 1 and 8 μg/mL and the value of 2 μg/mL was found for the majority of strains (49 isolates). Furthermore, Flamm et al. conducted a research on the activity of pexiganan against pathogens isolated from diabetic foot infection (DFI). [39]. The study involved not only bacteriological assays, but also characterization of resistance genes. The MICs for two DFI-associated A. baumannii strains were equal to 8 μg/mL. Moreover, pexiganan and colistin were also applied in experimental mouse models of A. baumannii infection [40]. In that study, the compounds were used for susceptibility determination and survival assessment of mice. It should be noticed that pexiganan was found to be effective in combinatory approach (low bacterial count) and also increased NK cytotoxic activity over the levels of infected

and colistin-treated animals. LL-37 is the third most active peptide assessed in that study and, at the same time, the most extensively studied one against A. baumannii [41–44]. In the research conducted by Feng et al., antibacterial and antibiofilm activities of LL-37 and its fragments (KS-30, KR-20, KR-12) against clinical strains were determined [41]. Furthermore, not only MICs but also the kinetics, impact on biofilm formation, development, and dispersal were assessed. Although the MICs for the clinical strains applied in their study were higher than those obtained in this research (16 and 32 μg/mL versus 4 μg/mL), the impact on biofilm formation occurred in similar concentrations. Our study revealed that LL-37 eradicates biofilm at a very high concentration (MBEC = 512 μg/mL). This suggests that application of this peptide is much more recommendable for prevention of biofilm-associated infections than for their treatment. Aurein 1.2 and citropin 1.1 are the peptides of amphibian origin that in our study exhibited identical activities against planktonic forms of A. baumannii (16 μg/mL) and the biofilm on tracheal tubes (32 μg/mL). It should be emphasized that the MBEC value obtained on polystyrene plates was four times lower for aurein 1.2 (256 versus 64 μg/mL). Despite the fact those peptides were extensively studied for their antimicrobial and physicochemical properties, their impact on A. baumannii has not been [45–50]. Only aurein 1.2, in a study conducted by de Freitas et al. on the enhancement of antimicrobial photodynamic therapy, has been found to reduce significantly the viability of S. aureus, Escherichia coli, and A. baumannii as well [51]. Omiganan and its retro analog (r-omiganan) were also tested in our study; however, only the first one has been studied against A. baumannii so far. In a study conducted by Sader et al., the antimicrobial activity of omiganan pentachloride (formerly MBI 226) was determined against more than one and a half thousand clinical isolates of bacteria and yeasts [52]. Moreover, not only the susceptibility profiles to antimicrobial peptide and conventional antibiotics was assessed, but also killing kinetics and precise quantification of resistant strains were estimated. The MIC obtained with the strain A. baumannii 101-2823A for omiganan was 8 μg/mL, which is twice lower than that determined in our study. In addition, r-omiganan was found to be a stronger antimicrobial than the parent compound. The results are consistent with our previous work where the retro analog of omiganan exhibited improved activity against Gram-negative strains as compared to that of the parent peptide. At the same time, omiganan is less cytotoxic than its retro counterpart owing to the lower rate of hemolysis. [53]. Temporin A was the weakest peptide tested in our study and the most cytotoxic one. The activity against planktonic forms was 128 μg/mL, which was 16 times higher than the IC50. Even more, the MBEC values for tracheal tube fragments and polystyrene were twice and four times higher, respectively. As a matter of fact, temporin A was previously tested against A. baumannii. In a study by Mangoni et al.,

Probiotics & Antimicro. Prot.

temporins A, B, and G were tested against multidrug-resistant clinical isolates of multiple bacterial strains involved in nosocomial infections. The results reported by this group indicated that the A. baumannii strains were sensitive to temporins over a concentration range comparable or lower than that required to kill Gram-positive strains such as S. aureus. However, these results are inconsistent with our findings, due to the different protocols of antimicrobial assay as well as the use of human serum in antimicrobial determination. Selectivity indexes (Table 3) calculated for all peptides suggest that citropin 1.1, pexiganan, and temporin A are distinctly cytotoxic (HaCaT) and MIC is relatively high (SI between 0.06 and 0.52). The highest SIs were determined for LL-37 and omiganan (2.93 and 2.48, respectively). Moreover, retro omiganan and CAMEL have slightly lower SIs (1.84 and 1.66, respectively). Nevertheless, different methods, particularly the materials, used for determination of antimicrobial activity can also give inconsistent results. This should be highlighted that positively charged molecules like AMPs may bind to the polystyrene surface. In our study, the polystyrene plates were used for determination of MIC and MBEC as well. For instance, Giacometti et al. have compared the activity of eight AMPs using two protocols, the first described by CLSI (formerly NCCLS) and the second by R.E.W. Hancock, which utilizes the use of polypropylene instead [54]. The results highlighted significant differences between those methods and indicated the higher activity of AMPs when the polypropylene plate was used. However, opposite observations were found in the study of Sánchez-Gómez et al. [55]. Therefore, it is reasoned to evaluate the activity of AMPs against A. baumannii on polystyrene and polypropylene plates in further studies.

Conclusions Despite the fact that the peptides used in this study, except temporin A, were found to be effective against planktonic forms of the reference strain A. baumannii ATCC 19606, their activity against biofilm formed on polystyrene and tracheal tube fragments was distinctly lower, especially as compared to the activities of conventional antibiotics. Nevertheless, it should be emphasized that the main characteristic of Acinetobacter spp. is their capability to acquire resistance. In fact, there is a risk that the compounds being effective against the reference strains may be inapplicable clinically. For this reason, the therapy of many infections caused by these bacteria is so challenging. Our study revealed how difficult is to eradicate the A. baumannii biofilm and indicated the difference between the characters of the surface where it has been formed. The peptides tested in our study turned out to be weaker antimicrobials than antibiotics. However, it should be noted that peptides as positively charged molecules may bind to the surface of polystyrene and therefore, their

antimicrobial activity may be underestimated. This notwithstanding, the compounds constitute a suitable basis for the design of new compounds with improved activity. Moreover, they proved to be more effective in combinatory approach. Many studies have shown their capability of binding to indwelling devices, thus making them resistant to biofilm formation. That is why the research on this particular group should be continued, with the focus on their biological and physicochemical characterization. Acknowledgements We wish to thank Professor Ryszard Piękoś for his invaluable help in preparing the manuscript. Author Contributions The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript. Funding This study was supported by the Medical University of Gdansk statutory grant No. ST-02-0087/07/508 and the Polish Ministry of Science and Higher Education Grant for Young Investigators, No. 010305/08/508.

Compliance with Ethical Standards Conflict of Interest The authors declare that they have no competing interests. Research Involving Human Participants and/or Animals The article does not contain any studies in patients by any of the authors. This article does not contain studies with human or animal subjects performed by any of the authors that should be approved by Ethics Committee. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http:// creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

References 1.

Almasaudi SB (2018) Acinetobacter spp. as nosocomial pathogens: epidemiology and resistance features. Saudi J Biol Sci 25:586–596. https://doi.org/10.1016/J.SJBS.2016.02.009 2. Appleman MD, Belzberg H, Citron DM, et al (2000) In vitro activities of nontraditional antimicrobials against multiresistant acinetobacter baumannii strains isolated in an intensive care unit outbreak. 44:1035–1040 3. Nemec A, Krizova L, Maixnerova M, van der Reijden TJK, Deschaght P, Passet V, Vaneechoutte M, Brisse S, Dijkshoorn L (2011) Genotypic and phenotypic characterization of the Acinetobacter calcoaceticus–Acinetobacter baumannii complex with the proposal of Acinetobacter pittii sp. nov. (formerly Acinetobacter genomic species 3) and Acinetobacter nosocomialis sp. nov. (formerly Acinetobacter genomic species 13TU). Res Microbiol 162:393–404. https://doi.org/10.1016/J.RESMIC.2011. 02.006

Probiotics & Antimicro. Prot. 4.

5.

6.

7.

8.

9.

10.

11.

12.

13. 14.

15.

16. 17.

18.

19.

20.

21.

22.

Ramette A, Kronenberg A (2018) Prevalence of carbapenemresistant Acinetobacter baumannii from 2005 to 2016 in Switzerland. BMC Infect Dis 18:159. https://doi.org/10.1186/ s12879-018-3061-5 Yang Y-S, Lee Y-T, Huang T-W, Sun JR, Kuo SC, Yang CH, Chen TL, Lin JC, Fung CP, Chang FY (2013) Acinetobacter baumannii nosocomial pneumonia: is the outcome more favorable in nonventilated than ventilated patients? BMC Infect Dis 13:142. https://doi.org/10.1186/1471-2334-13-142 Joly-Guillou M-L (2005) Clinical impact and pathogenicity of Acinetobacter. Clin Microbiol Infect 11:868–873. https://doi.org/ 10.1111/J.1469-0691.2005.01227.X Singh RK, Bhoi SK, Kalita J, Misra UK (2017) Multidrug-resistant Acinetobacter meningitis treated by intrathecal colistin. Ann Indian Acad Neurol 20:74–75. https://doi.org/10.4103/0972-2327.199908 Doughari HJ, Ndakidemi PA, Human IS, Benade S (2011) The ecology, biology and pathogenesis of Acinetobacter spp.: an overview. Microbes Environ 26:101–112. https://doi.org/10.1264/ jsme2.ME10179 Fan B, Guan J, Wang X, Cong Y (2016) Activity of colistin in combination with meropenem, tigecycline, fosfomycin, fusidic acid, rifampin or sulbactam against extensively drug-resistant Acinetobacter baumannii in a murine thigh-infection model. PLoS One 11:e0157757. https://doi.org/10.1371/journal.pone.0157757 Kim W-Y, Moon J-Y, Huh JW, Choi SH, Lim CM, Koh Y, Chong YP, Hong SB (2016) Comparable efficacy of tigecycline versus colistin therapy for multidrug-resistant and extensively drugresistant Acinetobacter baumannii pneumonia in critically ill patients. PLoS One 11:e0150642. https://doi.org/10.1371/journal. pone.0150642 Rao GG, Ly NS, Diep J, Forrest A, Bulitta JB, Holden PN, Nation RL, Li J, Tsuji BT (2016) Combinatorial pharmacodynamics of polymyxin B and tigecycline against heteroresistant Acinetobacter baumannii. Int J Antimicrob Agents 48:331–336. https://doi.org/10. 1016/j.ijantimicag.2016.06.006 Mansour SC, Pena OM, Hancock RE (2014) Host defense peptides: front-line immunomodulators. Trends Immunol 35:443–450. https://doi.org/10.1016/j.it.2014.07.004 Zasloff M (2002) Antimicrobial peptides of multicellular organisms. Nature 415:389–395. https://doi.org/10.1038/415389a Boman HG (2003) Antibacterial peptides: basic facts and emerging concepts. J Intern Med 254:197–215. https://doi.org/10.1046/j. 1365-2796.2003.01228.x Schauber J, Gallo RL (2007) Expanding the roles of antimicrobial peptides in skin: alarming and arming keratinocytes. J Invest Dermatol 127:510–512. https://doi.org/10.1038/sj.jid.5700761 Kamysz W, Okrój M, Łukasiak J (2003) Novel properties of antimicrobial peptides. Acta Biochim Pol 50:461–469 Fan L, Sun J, Zhou M, Zhou J, Lao X, Zheng H, Xu H (2016) DRAMP: a comprehensive data repository of antimicrobial peptides. Sci Rep 6:24482. https://doi.org/10.1038/srep24482 Hancock RE, Lehrer R (1998) Cationic peptides: a new source of antibiotics. Trends Biotechnol 16:82–88. https://doi.org/10.1016/ S0167-7799(97)01156-6 Govender T, Dawood A, Esterhuyse AJ, Katerere DR (2012) Antimicrobial properties of the skin secretions of frogs. S Afr J Sci 108. https://doi.org/10.4102/sajs.v108i5/6.795 Clinical and Laboratory Standards Institute (CLSI) (2012) Methods for dilution antimicrobial susceptibility tests f or bacteria that grow aerobically; approved standard—Ninth Edition Sikora K, Jaśkiewicz M, Neubauer D, et al (2018) Counter-ion effect on antistaphylococcal activity and cytotoxicity of selected antimicrobial peptides. Amino Acids 1–11. doi: 10.1007/s00726017-2536-9 Pfalzgraff A, Brandenburg K, Weindl G (2018) Antimicrobial peptides and their therapeutic potential for bacterial skin infections and

23. 24. 25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

wounds. Front Pharmacol 9:281. https://doi.org/10.3389/fphar. 2018.00281 Donlan RM (2002) Biofilms: microbial life on surfaces. Emerg Infect Dis 8:881–890. https://doi.org/10.3201/eid0809.020063 Mah TF, O’Toole GA (2001) Mechanisms of biofilm resistance to antimicrobial agents. Trends Microbiol 9:34–39 Jodoin J, Hincke MT (2018) Histone H5 is a potent antimicrobial agent and a template for novel antimicrobial peptides. Sci Rep 8: 2411. https://doi.org/10.1038/s41598-018-20912-1 Davies D (2003) Understanding biofilm resistance to antibacterial agents. Nat Rev Drug Discov 2:114–122. https://doi.org/10.1038/ nrd1008 Runci F, Bonchi C, Frangipani E, Visaggio D, Visca P (2017) Acinetobacter baumannii biofilm formation in human serum and disruption by gallium. Antimicrob Agents Chemother 61:e01563– e01516. https://doi.org/10.1128/AAC.01563-16 Dawgul M, Maciejewska M, Jaśkiewicz M et al (2014) Antimicrobial peptides as potential tool to fight bacterial biofilm. Acta Pol Pharm Drug Res 71:39–47 Maciejewska M, Bauer M, Neubauer D, Kamysz W, Dawgul M (2016) Influence of amphibian antimicrobial peptides and short lipopeptides on bacterial biofilms formed on contact lenses. Materials (Basel) 9:873. https://doi.org/10.3390/ma9110873 Dawgul M, Barańska-Rybak W, Bielińska S et al (2010) The influence of antimicrobial peptides on Candida biofilm. Alerg Astma Immunol 15:220–225 Wang K, Yan J, Dang W, Xie J, Yan B, Yan W, Sun M, Zhang B, Ma M, Zhao Y, Jia F, Zhu R, Chen W, Wang R (2014) Dual antifungal properties of cationic antimicrobial peptides polybia-MPI: membrane integrity disruption and inhibition of biofilm formation. Peptides 56:22–29. https://doi.org/10.1016/j.peptides.2014.03.005 Dean SN, Bishop BM, van Hoek ML (2011) Susceptibility of Pseudomonas aeruginosa biofilm to alpha-helical peptides: Denantiomer of LL-37. Front Microbiol 2:128. https://doi.org/10. 3389/fmicb.2011.00128 Machado MC, Webster TJ (2016) Decreased Pseudomonas aeruginosa biofilm formation on nanomodified endotracheal tubes: a dynamic lung model. Int J Nanomedicine 11:3825–3831. https:// doi.org/10.2147/IJN.S108253 Gil-Perotin S, Ramirez P, Marti V, Sahuquillo JM, Gonzalez E, Calleja I, Menendez R, Bonastre J (2012) Implications of endotracheal tube biofilm in ventilator-associated pneumonia response: a state of concept. Crit Care 16:R93. https://doi.org/10.1186/cc11357 Mulla S, Kumar A, Rajdev S (2016) Comparison of MIC with MBEC assay for in vitro antimicrobial susceptibility testing in biofilm forming clinical bacterial isolates. Adv Microbiol 06:73–78. https://doi.org/10.4236/aim.2016.62007 Wang Y-C, Huang T-W, Yang Y-S, et al Biofilm formation is not associated with worse outcome in Acinetobacter baumannii bacteraemic pneumonia. doi: 10.1038/s41598-018-25661-9 Inglis TJJ, Millar MR, Jones JG, Robinson4 DA (1989) Tracheal tube biofilm as a source of bacterial colonization of the lung. J Clin Microbiol 2014–2018 Giacometti A, Cirioni O, Kamysz W, D’Amato G, Silvestri C, del Prete MS, Łukasiak J, Scalise G (2003) Comparative activities of cecropin A, melittin, and cecropin A–melittin peptide CA(1– 7)M(2–9)NH2 against multidrug-resistant nosocomial isolates of Acinetobacter baumannii. Peptides 24:1315–1318. https://doi.org/ 10.1016/J.PEPTIDES.2003.08.003 Ge Y, Macdonald DL, Holroyd KJ et al (1999) In vitro antibacterial properties of pexiganan, an analog of magainin. Antimicrob Agents Chemother 43:782–788 Cirioni O, Simonetti O, Pierpaoli E, Barucca A, Ghiselli R, Orlando F, Pelloni M, Minardi D, Trombettoni MMC, Guerrieri M, Offidani A, Giacometti A, Provinciali M (2016) Enhanced efficacy of combinations of pexiganan with colistin versus Acinetobacter

Probiotics & Antimicro. Prot. baumannii in experimental sepsis. Shock 46:1–8. https://doi.org/10. 1097/SHK.0000000000000584 41. Feng X, Sambanthamoorthy K, Palys T, Paranavitana C (2013) The human antimicrobial peptide LL-37 and its fragments possess both antimicrobial and antibiofilm activities against multidrug-resistant Acinetobacter baumannii. Peptides 49:131–137. https://doi.org/10. 1016/J.PEPTIDES.2013.09.007 42. Babikir IH, Abugroun EA, Bilal NE, Alghasham AA, Abdalla EE, Adam I (2018) The impact of cathelicidin, the human antimicrobial peptide LL-37 in urinary tract infections. BMC Infect Dis 18:17. https://doi.org/10.1186/s12879-017-2901-z 43. Lin M-F, Tsai P-W, Chen J-Y, Lin YY, Lan CY (2015) OmpA binding mediates the effect of antimicrobial peptide LL-37 on Acinetobacter baumannii. PLoS One 10:e0141107. https://doi.org/ 10.1371/journal.pone.0141107 44. Mishra B, Wang G (2017) Titanium surfaces immobilized with the major antimicrobial fragment FK-16 of human cathelicidin LL-37 are potent against multiple antibiotic-resistant bacteria. Biofouling 33:544–555. https://doi.org/10.1080/08927014.2017.1332186 45. Maciejewska M, Bauer M, Neubauer D, Kamysz W, Dawgul M (2016) Influence of amphibian antimicrobial peptides and short lipopeptides on bacterial biofilms formed on contact lenses. Mater (Basel, Switzerland) 9: doi: 10.3390/ma9110873 46. Jorge P, Grzywacz D, Kamysz W, Lourenço A, Pereira MO (2017) Searching for new strategies against biofilm infections: colistinAMP combinations against Pseudomonas aeruginosa and Staphylococcus aureus single- and double-species biofilms. PLoS One 12:e0174654. https://doi.org/10.1371/journal.pone.0174654 47. Marcotte I, Wegener KL, Lam Y-H, Chia BCS, de Planque MRR, Bowie JH, Auger M, Separovic F (2003) Interaction of antimicrobial peptides from Australian amphibians with lipid membranes. Chem Phys Lipids 122:107–120. https://doi.org/10.1016/S00093084(02)00182-2 48. Dawgul M, Baranska-Rybak W, Piechowicz L, et al (2016) The antistaphylococcal activity of citropin 1.1 and temporin A against

planktonic cells and biofilms formed by isolates from patients with atopic dermatitis: an assessment of their potential to induce microbial resistance compared to conventional antimicrobials. Pharmaceuticals (Basel) 9. doi: 10.3390/ph9020030 49. Sikora K, Neubauer D, Jaśkiewicz M, Kamysz W (2017) Citropin 1.1 trifluoroacetate to chloride counter-ion exchange in HClsaturated organic solutions: an alternative approach. Int J Pept Res Ther 24:265–270. https://doi.org/10.1007/s10989-017-9611-7 50. Giacometti A, Cirioni O, Riva A, Kamysz W, Silvestri C, Nadolski P, Della Vittoria A, Lukasiak J, Scalise G (2007) In vitro activity of aurein 1.2 alone and in combination with antibiotics against grampositive nosocomial cocci. Antimicrob Agents Chemother 51: 1494–1496. https://doi.org/10.1128/AAC.00666-06 51. Marise De Freitas L, Lorenzón EN, Santos-Filho NA, et al Antimicrobial photodynamic therapy enhanced by the peptide aurein 1.2. doi: 10.1038/s41598-018-22687-x 52. Sader HS, Fedler KA, Rennie RP, Stevens S, Jones RN (2004) Omiganan pentahydrochloride (MBI 226), a topical 12-aminoacid cationic peptide: spectrum of antimicrobial activity and measurements of bactericidal activity. Antimicrob Agents Chemother 48:3112–3118. https://doi.org/10.1128/AAC.48.8.3112-3118.2004 53. Neubauer D, Jaśkiewicz M, Migoń D, Bauer M, Sikora K, Sikorska E, Kamysz E, Kamysz W (2017) Retro analog concept: comparative study on physico-chemical and biological properties of selected antimicrobial peptides. Amino Acids 49:1755–1771. https://doi. org/10.1007/s00726-017-2473-7 54. Giacometti A, Cirioni O, Barchiesi F, et al (2000) In vitro susceptibility tests for cationic peptides: comparison of broth microdilution methods for bacteria that grow aerobically. 44: 1694–1696 55. Sánchez-Gómez S, Lamata M, Leiva J, et al (2008) Comparative analysis of selected methods for the assessment of antimicrobial and membrane-permeabilizing activity: a case study for lactoferricin derived peptides. BMC Microbiol 8. doi: 10.1186/ 1471-2180-8-196